This review critically examines the current landscape of protein biophysics, highlighting a significant epistemological divide emerging from the rapid advancement of predictive algorithms compared to the slower pace of experimental validation. While artificial intelligence models and inferential statistical methods can generate millions of in silico protein interactions, experimental biophysics remains limited by physiological and technical challenges, leading to a scarcity of confirmed, functional data. This imbalance creates an environment where hypotheses and large-scale interactome networks proliferate based on unverified interactions. This fosters a false sense of causal understanding, potential misdirection in therapeutic development, and overinterpreting omics datasets. The challenge becomes even more urgent with complex, multifactorial diseases such as cancer and viral infections. Recognizing this subtle but crucial disparity is essential for researchers in protein biophysics to navigate the evolving landscape effectively and to focus validation efforts that support meaningful scientific and clinical progress. The review emphasizes the critical need for rigorous experimental validation alongside rapid computational advances. It highlights key challenges, including complexities in protein folding, dynamics, membrane interactions, and quantum effects. It discusses emerging technologies such as AI-based structure prediction (e.g., AlphaFold), cryo-electron microscopy, and integrated biophysical approaches. The importance of multidisciplinary collaboration and orthogonal validation methods is underscored to enhance the reliability of protein interaction data. While technological advances promise to deepen our understanding of protein functions and their roles in health and disease, the review advocates for cautious integration of predictive models with meticulous experimental verification, ensuring the development of accurate, biologically meaningful insights poised to advance medicine and biotechnology.
Citation: Giovanni Colonna. Protein biophysics: current limitations and prospects[J]. AIMS Biophysics, 2025, 12(3): 333-374. doi: 10.3934/biophy.2025018
This review critically examines the current landscape of protein biophysics, highlighting a significant epistemological divide emerging from the rapid advancement of predictive algorithms compared to the slower pace of experimental validation. While artificial intelligence models and inferential statistical methods can generate millions of in silico protein interactions, experimental biophysics remains limited by physiological and technical challenges, leading to a scarcity of confirmed, functional data. This imbalance creates an environment where hypotheses and large-scale interactome networks proliferate based on unverified interactions. This fosters a false sense of causal understanding, potential misdirection in therapeutic development, and overinterpreting omics datasets. The challenge becomes even more urgent with complex, multifactorial diseases such as cancer and viral infections. Recognizing this subtle but crucial disparity is essential for researchers in protein biophysics to navigate the evolving landscape effectively and to focus validation efforts that support meaningful scientific and clinical progress. The review emphasizes the critical need for rigorous experimental validation alongside rapid computational advances. It highlights key challenges, including complexities in protein folding, dynamics, membrane interactions, and quantum effects. It discusses emerging technologies such as AI-based structure prediction (e.g., AlphaFold), cryo-electron microscopy, and integrated biophysical approaches. The importance of multidisciplinary collaboration and orthogonal validation methods is underscored to enhance the reliability of protein interaction data. While technological advances promise to deepen our understanding of protein functions and their roles in health and disease, the review advocates for cautious integration of predictive models with meticulous experimental verification, ensuring the development of accurate, biologically meaningful insights poised to advance medicine and biotechnology.
| [1] |
Hille B (2022) A Life of Biophysics. Annu Rev Biophys 51: 1-17. https://doi.org/10.1146/annurev-biophys-120121-074034
|
| [2] |
Dill KA, Ozkan SB, Weikl TR (2007) The protein folding problem: when will it be solved?. Curr Opin Struc Biol 17: 342-346. https://doi.org/10.1016/j.sbi.2007.06.001
|
| [3] |
Murugan A, Husain K, Rust MJ, et al. (2021) Roadmap on biology in time varying environments. Phys Biol 18: 041502. https://doi.org/10.1088/1478-3975/abde8d
|
| [4] |
Bustin SA, Jellinger KA (2023) Advances in molecular medicine: unravelling disease complexity and pioneering precision healthcare. Int J Mol Sci 24: 14168. https://doi.org/10.3390/ijms241814168
|
| [5] |
Sharma A, Colonna G (2021) System-Wide Pollution of Biomedical Data: Consequence of the Search for Hub Genes of Hepatocellular Carcinoma Without Spatiotemporal Consideration. Mol Diagn Ther 25: 9-27. https://doi.org/10.1007/s40291-020-00505-3
|
| [6] |
Chautard E, Thierry-Mieg N, Ricard-Blum S (2009) Interaction networks: from protein functions to drug discovery. a review. Pathol Biol 57: 324-333. https://doi.org/10.1016/j.patbio.2008.10.004
|
| [7] |
Aebersold R, Agar JN, Amster IJ, et al. (2018) How many human proteoforms are there?. Nat Chem Biol 14: 206-214. https://doi.org/10.1038/nchembio.2576
|
| [8] |
Naryzhny S (2024) Puzzle of proteoform variety—Where is a key?. Proteomes 12: 15. https://doi.org/10.3390/proteomes12020015
|
| [9] |
Ashrafuzzaman M (2023) Introduction to Modern Biophysics. CRC Press. https://doi.org/10.1201/9781003287780
|
| [10] |
Lukyanenko R, Storey VC, Pastor O (2022) Conceptual modelling for life sciences based on systemist foundations. BMC bioinformatics 23: 574. https://doi.org/10.1186/s12859-023-05287-z
|
| [11] |
Wang X, Fan J (2021) Spatiotemporal molecular medicine: a new era of clinical and translational medicine. Clinical and Translational Medicine 11: e294. https://doi.org/10.1002/ctm2.294
|
| [12] |
Nakajima T (2013) Probability in biology: overview of a comprehensive theory of probability in living systems. Prog Biophys Mol Biol 113: 67-79. https://doi.org/10.1016/j.pbiomolbio.2013.03.007
|
| [13] |
Saeed N, Nam H, Haq M (2018) A survey on multidimensional scaling. ACM Comput Surv 51: 1-25. https://doi.org/10.1145/3178155
|
| [14] |
Buxbaum JN (2003) Diseases of protein conformation: what do in vitro experiments tell us about in vivo diseases?. Trends in biochemical sciences 28: 585-592. https://doi.org/10.1016/j.tibs.2003.09.009
|
| [15] |
Mazal H, Haran G (2019) Single-molecule FRET methods to study the dynamics of proteins at work. Curr Opin Biomed Eng 12: 8-17. https://doi.org/10.1016/j.cobme.2019.08.007
|
| [16] |
Uchihashi T, Scheuring S (2018) Applications of high-speed atomic force microscopy to real-time visualization of dynamic biomolecular processes. BBA-Gen Subjects 1862: 229-240. https://doi.org/10.1016/j.bbagen.2017.07.010
|
| [17] |
Ando T, Fukuda S, Ngo KX, et al. (2024) High-speed atomic force microscopy for filming protein molecules in dynamic action. Annu Rev Biophys 53: 19-39. https://doi.org/10.1146/annurev-biophys-030722-113353
|
| [18] |
Karunanithy G, Shukla VK, Hansen DF (2021) Methodological advancements for characterising protein side chains by NMR spectroscopy. Curr Opin Struct Biol 70: 61-69. https://doi.org/10.1016/j.sbi.2021.04.002
|
| [19] |
Huggins DJ, Biggin PC, Dämgen MA, et al. (2019) Biomolecular simulations: from dynamics and mechanisms to computational assays of biological activity. Wires: Comput Mol Sci 9: e1393. https://doi.org/10.1002/wcms.1393
|
| [20] |
Nelson LK (2020) Computational grounded theory: a methodological framework. Sociol Method Res 49: 3-42. https://doi.org/10.1177/0049124117729703
|
| [21] |
Monteiro da Silva G, Cui JY, Dalgarno DC, et al. (2024) High-throughput prediction of protein conformational distributions with subsampled AlphaFold2. Nat Commun 15: 2464. https://doi.org/10.1038/s41467-024-46715-9
|
| [22] |
Sadybekov AV, Katritch V (2023) Computational approaches streamlining drug discovery. Nature 616: 673-685. https://doi.org/10.1038/s41586-023-05905-z
|
| [23] |
Wang M, Ma A, Wang H, et al. (2024) Atomic molecular dynamics simulation advances of de novo-designed proteins. Q Rev Biophys 57: e14. https://doi.org/10.1017/S0033583524000131
|
| [24] |
Ose NJ, Campitelli P, Modi T, et al. (2024) Some mechanistic underpinnings of molecular adaptations of SARS-COV-2 spike protein by integrating candidate adaptive polymorphisms with protein dynamics. eLife 12: RP92063. https://doi.org/10.7554/eLife.92063.3
|
| [25] |
Fabbrizi E, Fiorentino F, Casano F, et al. (2025) Native mass spectrometry for proximity-inducing compounds: a new opportunity for studying chemical-induced protein modulation. Expert Opin Drug Dis 20: 643-657. https://doi.org/10.1080/17460441.2025.2486146
|
| [26] | Helmerhorst E, Chandler DJ, Nussio M, et al. (2012) Real-time and label-free bio-sensing of molecular interactions by surface plasmon resonance: a laboratory medicine perspective. Clin Biochem Rev 33: 161-173. |
| [27] |
Cavalcanti IDL, Junior FHX, Magalhães NSS, et al. (2023) Isothermal titration calorimetry (ITC) as a promising tool in pharmaceutical nanotechnology. Int J Pharm 641: 123063. https://doi.org/10.1016/j.ijpharm.2023.123063
|
| [28] |
dos Santos Rodrigues FH, Delgado GG, da Costa TS, et al. (2023) Applications of fluorescence spectroscopy in protein conformational changes and intermolecular contacts. BBA Adv 3: 100091. https://doi.org/10.1016/j.bbadva.2023.100091
|
| [29] |
AlQuraishi M, Sorger PK (2021) Differentiable biology: using deep learning for biophysics-based and data-driven modeling of molecular mechanisms. Nat Methods 18: 1169-1180. https://doi.org/10.1038/s41592-021-01283-4
|
| [30] |
Gong R, Reynolds MJ, Carney KR, et al. (2025) Fascin structural plasticity mediates flexible actin bundle construction. Nat Struct Mol Biol 32: 940-952. https://doi.org/10.1038/s41594-024-01477-2
|
| [31] |
Piper SJ, Johnson RM, Wootten D, et al. (2022) Membranes under the magnetic lens: a dive into the diverse world of membrane protein structures using Cryo-EM. Chem Rev 122: 13989-14017. https://doi.org/10.1021/acs.chemrev.1c00837
|
| [32] | Duckhoe K, Sahin O (2015) Imaging and three-dimensional reconstruction of chemical groups inside a protein complex using atomic force microscopy. Nat Nanotechnol 10.3: 264-269. https://doi.org/10.1038/nnano.2014.335 |
| [33] |
Uzorka A (2025) Single-molecule biophysics: probing biological processes at the nanoscale. Biophys Rev Lett 20: 13-29. https://doi.org/10.1142/S1793048025300014
|
| [34] |
Sonar R, Mishra PP (2025) Exploring dynamics at the mmolecular sscale: advances in single-molecule biophysics. Biochemical and Biophysical Methods in Molecular and Cellular Biology . Singapore: Springer Nature Singapore 163-184. https://doi.org/10.1007/978-981-96-2088-3_7
|
| [35] |
Miller H, Zhou Z, Shepherd J, et al. (2017) Single-molecule techniques in biophysics: a review of the progress in methods and applications. Rep Prog Phys 81: 024601. https://doi.org/10.48550/arXiv.1704.06837
|
| [36] | Zhang L, Shen D, Yang J (2025) Advancing Single-Molecule Biophysics: Next-Generation Organic Fluorophores with Tailored Labeling Strategies. Chem Biomed Imag . https://doi.org/10.1021/cbmi.5c00007 |
| [37] |
Czajkowsky DM, Sun J, Shen Y, et al. (2015) Single molecule compression reveals intra-protein forces drive cytotoxin pore formation. elife 4: e08421. https://doi.org/10.7554/eLife.08421
|
| [38] |
Kopac T (2025) Leveraging artificial intelligence and machine learning for characterizing protein corona, nanobiological interactions, and advancing drug discovery. Bioengineering 12: 312. https://doi.org/10.3390/bioengineering12030312
|
| [39] |
Jumper J, Evans R, Pritzel A, et al. (2021) Highly accurate protein structure prediction with AlphaFold. Nature 596: 583-589. https://doi.org/10.1038/s41586-021-03819-2
|
| [40] |
Abramson J, Adler J, Dunger J, et al. (2024) Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature 630: 493-500. https://doi.org/10.1038/s41586-024-07487-w
|
| [41] |
Brotzakis ZF, Zhang S, Murtada MH, et al. (2025) AlphaFold prediction of structural ensembles of disordered proteins. Nat Commun 16: 1632. https://doi.org/10.1038/s41467-025-56572-9
|
| [42] |
Ruffolo JA, Chu LS, Mahajan SP, et al. (2023) Fast, accurate antibody structure prediction from deep learning on massive set of natural antibodies. Nat Commun 14: 2389. https://doi.org/10.1038/s41467-023-38063-x
|
| [43] |
Gasparyan L, Mazo I, Simonyan V, et al. (2019) DNA Sequencing: current state and prospects of development. Open J Biophys 9: 169-197. https://doi.org/10.4236/ojbiphy.2019.93013
|
| [44] |
Monteiro da Silva G, Cui JY, Dalgarno DC, et al. (2024) High-throughput prediction of protein conformational distributions with subsampled AlphaFold2. Nat Commun 15: 2464. https://doi.org/10.1038/s41467-024-46715-9
|
| [45] |
Liu H, Zhuo C, Gao J, et al. (2024) AI-integrated network for RNA complex structure and dynamic prediction. Biophys Rev 5: 041304. https://doi.org/10.1063/5.0237319
|
| [46] | Castaño-Díez D, Danev R, Baumeister W (2020) Progress in cryo-EM aided by new technological advances. Curr Opin Struct Biol 64: 37-48. https://doi.org/10.1016/j.sbi.2020.01.006 |
| [47] |
Lee M (2023) Recent advances in deep learning for protein-protein interaction analysis: a comprehensive review. Molecules 28: 5169. https://doi.org/10.3390/molecules28135169
|
| [48] |
Schauperl M, Denny RA (2022) AI-based protein structure prediction in drug discovery: impacts and challenges. J Chem Inf Model 62: 3142-3156. https://doi.org/10.1021/acs.jcim.2c00026
|
| [49] |
Nussinov R, Tsai CJ, Shehu A, et al. (2019) Computational structural biology: successes, future directions, and challenges. Molecules 24: 637. https://doi.org/10.3390/molecules24030637
|
| [50] |
Dobson CM (2019) Biophysical techniques in structural biology. Annu Rev Biochem 88: 25-33. https://doi.org/10.1146/annurev-biochem-013118-111947
|
| [51] |
Miller H, Zhou Z, Shepherd J, et al. (2017) Single-molecule techniques in biophysics: a review of the progress in methods and applications. Rep Prog Phys 81: 024601. https://iopscience.iop.org/article/10.1088/1361-6633/aa8a02
|
| [52] |
Zhang Z, Kumar V, Dybkov O, et al. (2024) Cryo-EM analyses of dimerized spliceosomes provide new insights into the functions of B complex proteins. EMBO Journal 43: 1065-1088. https://doi.org/10.1038/s44318-024-00052-1
|
| [53] |
Yeukuang H, Margaritondo G (2021) Synchrotron radiation and X-ray free-electron lasers (X-FELs) explained to all users, active and potential. Synchrotron Radiat 28: 1014-1029. https://doi.org/10.1107/S1600577521003325
|
| [54] |
Sperger T, Sanhueza IA, Schoenebeck F (2016) Computation and experiment: a powerful combination to understand and predict reactivities. Acc Chem Res 49: 1311-1319. https://doi.org/10.1021/acs.accounts.6b00068
|
| [55] |
Chang L, Mondal A, Singh B, et al. (2024) Revolutionizing peptide-based drug discovery: advances in the post-AlphaFold era. Wires: Comput Mol Sci 14: e1693. https://doi.org/10.1002/wcms.1693
|
| [56] |
Sonn-Segev A, Belacic K, Bodrug T, et al. (2020) Quantifying the heterogeneity of macromolecular machines by mass photometry. Nat Commun 11: 1772. https://doi.org/10.1038/s41467-020-15642-w
|
| [57] |
Rathore AS, Sarin D (2024) What should next-generation analytical platforms for biopharmaceutical production look like?. Trends Biotechnol 42: 282-292. https://doi.org/10.1016/j.tibtech.2023.08.008
|
| [58] |
Jeon H, Han J (2025) Microfluidics with machine learning for biophysical characterization of cells. Annu Rev Anal Chem 18: 447-472. https://doi.org/10.1146/annurev-anchem-061622-025021
|
| [59] |
Silva TC, Eppink M, Ottens M (2022) Automation and miniaturization: enabling tools for fast, high-throughput process development in integrated continuous biomanufacturing. J Chem Technol Biotechnol 97: 2365-2375. https://doi.org/10.1002/jctb.6792
|
| [60] | Krishnan M, Darling S (2020) Meeting analytical challenges in a brave new world of next generation biotherapeutics. Drug Discov 21: 31-39. |
| [61] |
Capelle MA, Gurny R, Arvinte T (2007) High throughput screening of protein formulation stability: practical considerations. Eur J Pharm Biopharma 65: 131-148. https://doi.org/10.1016/j.ejpb.2006.09.009
|
| [62] |
Kwan TO, Kolek SA, Danson AE, et al. (2022) Measuring protein aggregation and stability using high-throughput biophysical approaches. Front Mol Biosci 9: 890862. https://doi.org/10.3389/fmolb.2022.890862
|
| [63] |
Lindsey YN, Villa E (2023) Bringing structure to cell biology with cryo-electron tomography. Annu Rev Biophys 52: 573-595. https://doi.org/10.1146/annurev-biophys-111622-091327
|
| [64] |
Petoukhov MV, Svergun DI (2012) Small angle X-ray scattering/small angle neutron scattering as methods complementary to NMR. NMR of Biomolecules: Towards Mechanistic Systems Biology : 562-574. https://doi.org/10.1002/9783527644506.ch35
|
| [65] |
Ashkar R, Bilheux H, Bordallo H, et al. (2018) Neutron scattering in the biological sciences: progress and prospects. Acta Cryst Struct Biol 74: 1129-1168. https://doi.org/10.1107/S2059798318017503
|
| [66] |
Barbieri L, Luchinat E, Banci L (2016) Characterization of proteins by in-cell NMR spectroscopy in cultured mammalian cells. Nat Protoc 11: 1101-1111. https://doi.org/10.1038/nprot.2016.061
|
| [67] |
Serber Z, Corsini L, Durst F, et al. (2005) In-cell NMR spectroscopy. Method Enzymol 394: 17-41. https://doi.org/10.1016/S0076-6879(05)94002-0
|
| [68] |
Smith MJ, Marshall CB, Theillet FX, et al. (2015) Real-time NMR monitoring of biological activities in complex physiological environments. Curr Opin Struct Biol 32: 39-47. https://doi.org/10.1016/j.sbi.2015.02.003
|
| [69] |
Breindel L, Burz DS, Shekhtman A (2020) Active metabolism unmasks functional protein–protein interactions in real time in-cell NMR. Commun Biol 3: 249. https://doi.org/10.1038/s42003-020-0976-3
|
| [70] | Xiang X, Hansen AL, Bruschweiler-Li L, et al. (2025) Detection of intramolecular protein dynamics on nanosecond-to-microsecond timescales by nanoparticle-assisted NMR spin relaxation (NASR). Nat Protoc 434. https://doi.org/10.1038/s41596-025-01177-1 |
| [71] |
Novotny J, Komorovsky S, Marek R (2024) Paramagnetic effects in NMR spectroscopy of transition-metal complexes: principles and chemical concepts. Acc Chem Res 57: 1467-1477. https://doi.org/10.1021/acs.accounts.3c00786
|
| [72] | Schreier S, Polnaszek CF, Smith IC (1978) Spin labels in membranes problems in practice. BBA-Rev Biomemb 515: 395-436. https://doi.org/10.1016/0304-4157(78)90011-4 |
| [73] | Bren KL (2015) NMR analysis of spin densities. Spin States in Biochemistry and Inorganic Chemistry: Influence on Structure and Reactivity . Wiley 409-434. https://doi.org/10.1002/9781118898277.ch16 |
| [74] |
Masson GR, Jenkins ML, Burke JE (2017) An overview of hydrogen deuterium exchange mass spectrometry (HDX-MS) in drug discovery. Expert Opin Drug Dis 12: 981-994. https://doi.org/10.1080/17460441.2017.1363734
|
| [75] |
Damont A, Legrand A, Cao C, et al. (2023) Hydrogen/deuterium exchange mass spectrometry in the world of small molecules. Mass Spectrom Rev 42: 1300-1331. https://doi.org/10.1002/mas.21765
|
| [76] |
Lopez del Amo J, Uwe F, Bernd R (2010) Quantification of protein backbone hydrogen-deuterium exchange rates by solid state NMR spectroscopy. J Biomol NMR 48: 203-212. https://doi.org/10.1007/s10858-010-9450-8
|
| [77] |
Ziegler SJ, Mallinson SJB, St. John PC, et al. (2021) Advances in integrative structural biology: Towards understanding protein complexes in their cellular context. Comput Struct Biotechnol J 19: 214-225. https://doi.org/10.1016/j.csbj.2020.11.052
|
| [78] |
White SH (2023) Fifty years of biophysics at the membrane frontier. Annu Rev Biophys 52: 21-67. https://doi.org/10.1146/annurev-biophys-051622-112341
|
| [79] | Zhang L, Shen D, Yang J (2025) Advancing single-molecule biophysics: next-generation organic fluorophores with tailored labeling strategies. Chem Biomed Imag . https://doi.org/10.1021/cbmi.5c00007 |
| [80] |
D'Imprima E, Kühlbrandt W (2021) Current limitations to high-resolution structure determination by single-particle cryoEM. Quart Rev Biophys 54: 1-15. https://doi.org/10.1017/S003358352100002
|
| [81] |
Piper SJ, Johnson RM, Wootten D, et al. (2022) Membranes under the magnetic lens: a dive into the diverse world of membrane protein structures using Cryo-EM. Chem Rev 122: 13989-14017. https://doi.org/10.1021/acs.chemrev.1c00837
|
| [82] |
Yael P, Schreiber G (2013) Formation of protein complexes in crowded environments–from in vitro to in vivo. FEBS Lett 587: 1046-1052. https://doi.org/10.1016/j.febslet.2013.01.007
|
| [83] |
Vikulina AS, Skirtach AG, Volodkin D (2019) Hybrids of polymer multilayers, lipids, and nanoparticles: mimicking the cellular microenvironment. Langmuir 35: 8565-8573. https://doi.org/10.1021/acs.langmuir.8b04328
|
| [84] |
Moffat K (2025) Dynamics and kinetics in structural biology: the example of DNA photolyase. Quart Rev Biophys 58: 1-15. https://doi.org/10.1017/S0033583524000222
|
| [85] |
Dufourc EJ (2021) Bicelles and nanodiscs for biophysical chemistry. BBA-Biomembranes 1863: 183478. https://doi.org/10.1016/j.bbamem.2020.183478
|
| [86] |
Li H, Sun X, Cui W, et al. (2024) Computational drug development for membrane protein targets. Nat Biotechnol 42: 229-242. https://doi.org/10.1038/s41587-023-01987-2
|
| [87] |
Merz KM (2014) Using quantum mechanical approaches to study biological systems. Acc Chem Res 47: 2804-2811. https://doi.org/10.1021/ar5001023
|
| [88] |
Zoler E, Meyer T, Bellón JS, et al. (2024) Promiscuous Janus kinase binding to cytokine receptors modulates signaling efficiencies and contributes to cytokine pleiotropy. Sci Signal 17: eadl1892. https://doi.org/10.1126/scisignal.adl1892
|
| [89] |
Mucs D, Bryce RA (2013) The application of quantum mechanics in structure-based drug design. Expert Opin Drug Dis 8: 263-276. https://doi.org/10.1517/17460441.2013.752812
|
| [90] |
Sousa SF, Ribeiro AJ, Neves RP, et al. (2017) Application of quantum mechanics/molecular mechanics methods in the study of enzymatic reaction mechanisms. Wires: Comput Mol Sci 7: e1281. https://doi.org/10.1002/wcms.1281
|
| [91] |
Brinks D, Hildner R, van Dijk EMKP, et al. (2014) Ultrafast dynamics of single molecules. Chem Soc Rev 43: 2476-2491. https://doi.org/10.1039/C3CS60269A
|
| [92] | Sharples F, Bremer A, Moss SM, et al. (2021) Quantum science concepts in enhancing sensing and imaging technologies. In Applications for Biology: Proceedings of a Workshop. Technologies: Applications for Biology: Proceedings of a Workshop . Washington, DC: The National Academies Press. https://doi.org/10.17226/26139 |
| [93] |
Shubin Q (2025) Comparison of classical and hybrid quantum models in secondary structure protein prediction. Int J High School Res 7: 87-96. https://doi.org/10.36838/v7i3.14
|
| [94] |
Melkikh AV, Khrennikov A (2015) Nontrivial quantum and quantum-like effects in biosystems: Unsolved questions and paradoxes. Prog Biophys Mol Biol 119: 137-161. https://doi.org/10.1016/j.pbiomolbio.2015.07.001
|
| [95] | Aiello CD Quantum physics proposes a new way to study biology–and the results could revolutionize our understanding of how life works (2023). |
| [96] |
Ertelt M, Moretti R, Meiler J, et al. (2025) Self-supervised machine learning methods for protein design improve sampling but not the identification of high-fitness variants. Sci Adv 11: eadr7338. https://doi.org/10.1126/sciadv.adr7338
|
| [97] |
Dill KA, Ozkan SB, Weikl TR, et al. (2007) The protein folding problem: when will it be solved?. Curr Opin Struct Biol 7: 342-346. https://doi.org/10.1016/j.sbi.2007.06.001
|
| [98] |
Murugan A, Husain K, Rust MJ, et al. (2021) Roadmap on biology in time varying environments. Phys Biol 18: 041502. https://doi.org/10.1088/1478-3975/abde8d
|
| [99] |
Dill KA, Ozkan SB, Shell MS, et al. (2008) The protein folding problem. Annu Rev Biophys 37: 289-316. https://doi.org/10.1146/annurev.biophys.37.092707.153558
|
| [100] |
Hansen J, Jain AR, Nenov P, et al. (2024) From transcriptomics to digital twins of organ function. Front Cell Develop Biol 12: 1240384. https://doi.10.3389/fcell.2024.1240384
|
| [101] |
Balchin D, Hayer-Hartl M, Hartl FU (2016) In vivo aspects of protein folding and quality control. Science 353: aac4354. https://doi.org/10.1126/science.aac4354
|
| [102] |
Zwanzig R, Szabo A, Bagchi B (1992) Levinthal's paradox. P Natl A Sci 89: 20-22. https://doi.org/10.1016/S1359-0278(97)00067-9
|
| [103] | Wang L, Wen Z, Liu SW, et al. (2024) Overview of AlphaFold2 and breakthroughs in overcoming its limitations. Comput Biol Med 108620. https://doi.org/10.1016/j.compbiomed.2024.108620 |
| [104] |
David A, Islam S, Tankhilevich E, et al. (2022) The AlphaFold database of protein structures: a biologist's guide. J Mol Biol 434: 167336. https://doi.org/10.1016/j.jmb.2021.167336
|
| [105] |
Grandori R (2023) Protein structure and dynamics in the era of integrative structural biology. Front Biophys 1: 1219843. https://doi.org/10.3389/frbis.2023.1219843
|
| [106] | Castaño-Díez D, Danev R, Baumeister W (2020) Progress in cryo-EM aided by new technological advances. Curr Opin Struct Biol 64: 37-48. https://doi.org/10.1016/j.sbi.2020.01.006 |
| [107] |
Chua EY, Mendez JH, Rapp M, et al. (2022) Better, faster, cheaper: recent advances in cryo–electron microscopy. Annu Rev Biochem 91: 1-32. https://doi:10.1146/annurev-biochem-032620-110705
|
| [108] |
D'Imprima E, Kühlbrandt W (2021) Current limitations to high-resolution structure determination by single-particle cryoEM. Quart Rev Biophys 54: 1-15. https://doi.org/10.1017/S003358352100002
|
| [109] |
Danev R, Yanagisawa H, Kikkawa M (2019) Cryo-electron microscopy mmethodology: current aspects and future directions. Trends Biochem Sci 44: 837-848. https://doi.org/10.1016/j.tibs.2019.04.008
|
| [110] |
Wang HW, Wang JW (2017) How cryo-electron microscopy and X-ray crystallography complement each other. Protein Sci 26: 32-39. https://doi.org/10.1002/pro.3022
|
| [111] |
Marion D (2013) An introduction to biological NMR spectroscopy. Mol Cell Proteomics 12: 3006-3025. https://doi.org/10.1074/mcp.O113.030239
|
| [112] |
Sugiki T, Kobayashi N, Fujiwara T (2017) Modern technologies of solution nuclear magnetic resonance spectroscopy for three-dimensional structure determination of proteins open avenues for life scientists. Comput Struct Biotechnol J 15: 328-339. https://doi.org/10.1016/j.csbj.2017.04.001
|
| [113] |
Caffrey M (2009) Crystallizing membrane proteins for structure determination: use of lipidic mesophases. Annu Rev Biophys 38: 29-51. https://doi.org/10.1146/annurev.biophys.050708.133655
|
| [114] |
Vinothkumar KR, Henderson R (2010) Structures of membrane proteins. Q Rev Biophys 43: 65-158. https://doi.org/10.1017/S0033583510000041
|
| [115] |
Pörtner R (2023) Biopharmaceutical Manufacturing. Springer. https://doi.org/10.1007/978-3-031-45669-5
|
| [116] |
Zhou HX (2013) Influence of crowded cellular environments on protein folding, binding, and oligomerization: biological consequences and potentials of atomistic modeling. FEBS Lett 587: 1053-1061. https://doi.org/10.1016/j.febslet.2013.01.064
|
| [117] |
Singh DB, Tripathi T (2020) Frontiers in Protein Structure, Function, and Dynamics. Berlin: Springer. https://doi.org/10.1007/978-981-15-5530-5
|
| [118] |
Son A, Kim W, Park J, et al. (2024) Utilizing molecular dynamics simulations, machine learning, Cryo-EM, and NMR spectroscopy to predict and validate protein dynamics. Int J Mol Sci 25: 9725. https://doi.org/10.3390/ijms25179725
|
| [119] |
Voelz VA, Singh VR, Wedemeyer WJ, et al. (2010) Unfolded-state dynamics and structure of protein L characterized by simulation and experiment. J Am Chem Soc 132: 4702-4709. https://doi.org/10.1021/ja908369h
|
| [120] |
Kuznetsova IM, Turoverov KK, Uversky VN (2014) What macromolecular crowding can do to a protein. Int J Mol Sci 15: 23090-23140. https://doi.org/10.3390/ijms151223090
|
| [121] |
Xin F, Radivojac P (2012) Post-translational modifications induce significant yet not extreme changes to protein structure. Bioinformatics 28: 2905-2913. https://doi.org/10.1093/bioinformatics/bts541
|
| [122] |
Van Den Bedem H, Fraser JS (2015) Integrative, dynamic structural biology at atomic resolution—it's about time. Nat Methods 12: 307-318. https://doi.org/10.1038/nmeth.3324
|
| [123] |
Lerner E, Cordes T, Ingargiola A, et al. (2018) Toward dynamic structural biology: Two decades of single-molecule Förster resonance energy transfer. Science 359: eaan1133. https://doi.org/10.1126/science.aan1133
|
| [124] |
Varadi M, Tsenkov M, Velankar S (2025) Challenges in bridging the gap between protein structure prediction and functional interpretation. Proteins 93: 400-410. https://doi.org/10.1002/prot.26614
|
| [125] |
Harmalkar A, Lyskov S, Gray JJ (2024) Reliable protein-protein docking with AlphaFold, Rosetta, and replica-exchange. eLife 13: RP94029. https://doi.org/10.7554/eLife.94029.2
|
| [126] |
Wang L, Wen Z, Liu S, et al. (2024) Overview of AlphaFold2 and breakthroughs in overcoming its limitations. Comput Biol Med 176: 108620. https://doi.org/10.1016/j.compbiomed.2024.108620
|
| [127] |
Yuan R, Zhang J, Zhou J, et al. (2025) Recent progress and future challenges in structure-based protein-protein interaction prediction. Mol Ther 33: 2252-2268. https://doi.org/10.1016/j.ymthe.2025.04.003
|
| [128] | Li J, Liu Y (2023) Data quality, data sampling and data fitting: a tutorial guide for constructing full-dimensional accurate potential energy surfaces (PESs) of molecules and reactions. Machine Learning in Molecular Sciences . Cham: Springer 161-201. https://doi.org/10.1007/978-3-031-37196-7_6 |
| [129] |
Krokidis MG, Dimitrakopoulos GN, Vrahatis AG, et al. (2024) Challenges and limitations in computational prediction of protein misfolding in neurodegenerative diseases. Front Comput Neurosci 17: 1323182. https://doi.org/10.3389/fncom.2023.1323182
|
| [130] |
Wei G, Xi W, Nussinov R., et al. (2016) Protein ensembles: how does nature harness thermodynamic fluctuations for life? The diverse functional roles of conformational ensembles in the cell. Chem Rev 116: 6516-6551. https://doi.org/10.1021/acs.chemrev.5b00562
|
| [131] |
Beck M, Covino R, Hänelt I, et al. (2024) Understanding the cell: future views of structural biology. Cell 187: 545-562. https://doi.org/10.1016/j.cell.2023.12.017
|
| [132] |
Hribar-Lee B, Lukšič M (2024) Biophysical principles emerging from experiments on protein–protein association and aggregation. Annu Rev Biophys 53: 1-18. https://doi.org/10.1146/annurev-biophys-030722-111729
|
| [133] |
De Las Rivas J, Fontanillo C (2012) Protein–protein interaction networks: unraveling the wiring of molecular machines within the cell. Brief Funct Genomics 11: 489-496. https://doi.org/10.1093/bfgp/els036
|
| [134] |
Barabási AL, Bonabeau E (2003) Scale-free networks. Sci Am 288: 60-69. https://www.jstor.org/stable/26060284
|
| [135] |
Loscalzo J, Barabasi AL (2011) Systems biology and the future of medicine. Wires: Syst Biol Med 3: 619-627. https://doi.org/10.1002/wsbm.144
|
| [136] |
Goh KI, Cusick ME, Valle D, et al. (2007) The human disease networks. P Natl A Sci 104: 8685-8690. https://doi.org/10.1073/pnas.0701361104
|
| [137] |
Braun P, Gingras AC (2012) History of protein–protein interactions: from egg-white to complex networks. Proteomics 12: 1478-1498. https://doi.org/10.1002/pmic.201100563
|
| [138] |
Bajorath J (2002) Integration of virtual and high-throughput screening. Nat Rev Drug Discov 1: 882-894. https://doi.org/10.1038/nrd941
|
| [139] |
Jessulat M, Pitre S, Gui Y, et al. (2011) Recent advances in protein–protein interaction prediction: experimental and computational methods. Expert Opin Drug Dis 6: 921-935. https://doi.org/10.1517/17460441.2011.603722
|
| [140] |
Huttlin EL, Ting L, Bruckner RJ, et al. (2015) The BioPlex network: a systematic exploration of the human interactome. Cell 162: 425-440. https://doi.org/10.1016/j.cell.2015.06.043
|
| [141] |
Cizauskas C, DeBenedictis, Kelly P (2025) How the past is shaping the future of life science: The influence of automation and AI on biology. New Biotechnol 88: 1-11. https://doi.org/10.1016/j.nbt.2025.03.004
|
| [142] | Colonna G How digitalization and algorithms are changing the knowledge in biomedicine, preprints (2025). https://www.preprints.org/manuscript/202507.1086/v1 |
| [143] |
Gendler TS (1998) Galileo and the indispensability of scientific thought experiment. Brit J Philos Sci 49: 397-424. https://doi.org/10.1093/bjps/49.3.397
|
| [144] | Turing AM (2021) Computing machinery and intelligence (1950). Mind 59: 433-460. https://doi.org/10.1093/oso/9780198250791.003.0017 |
| [145] | Wu ST, Zhu L, Feng XL, et al. (2025) Strategies for discovering novel hepatocellular carcinoma biomarkers. World J Hepatol 17: 101201. http://dx.doi.org/10.4254/wjh.v17.i2.101201 |
| [146] |
Gao Q, Zhu H, Dong L, et al. (2019) Integrated proteogenomic characterization of HBV-related hepatocellular carcinoma. Cell 179: 561-577. https://doi.org/10.1016/j.cell.2019.08.052
|
| [147] | Costantini S, Capone F, Guerriero E, et al. (2010) Serum cytokine levels as putative prognostic markers in the progression of chronic HCV hepatitis to cirrhosis. Eur Cytokine Netw 21: 251-256. https://doi.org/10.1155/2015/707254 |
| [148] |
Capone F, Guerriero E, Colonna G, et al. (2014) Cytokinome profile evaluation in patients with hepatitis C virus infection. World J Gastroentero 20: 9261-9269. https://doi.org/10.3748/wjg.v20.i28.9261
|
| [149] |
Szklarczyk D, Gable AL, Nastou KC, et al. (2020) The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res 49: D605-D612. https://doi.org/10.1093/nar/gkaa1074
|
| [150] |
Szklarczyk D, Kirsch R, Koutrouli M, et al. (2022) The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res 51: D638-D646. https://doi.org/10.1093/nar/gkac1000
|
| [151] | Colonna G Unravelling the barriers: current limitations in cancer biology research and how to overcome them, preprints (2025). https://doi.org/10.20944/preprints202505.1837.v1 |
| [152] |
Deane CM, Salwiński Ł, Xenarios I, et al. (2002) Protein interactions: two methods for assessment of the reliability of high throughput observations. Mol Cell Proteom 11: 349-356. https://doi.org/10.1074/mcp.M100037-MCP200
|
| [153] |
Braun P (2012) Interactome mapping for analysis of complex phenotypes: Insights from benchmarking binary interaction assays. Proteomics 12: 1499-1518. https://doi.org/10.1002/pmic.201100598
|
| [154] |
Westermarck J, Ivaska J, Corthals GL (2013) Identification of Protein Interactions Involved in Cellular Signaling. Mol Cell Proteom 12: 1752. https://doi.org/10.1074/mcp.R113.027771
|
| [155] |
Brückner A, Polge C, Lentze N, et al. (2009) Yeast two-hybrid, a powerful tool for systems biology. Int J Mol Sci 10: 2763-2788. https://doi.org/10.3390/ijms10062763
|
| [156] |
Ito T, Chiba T, Ozawa R, et al. (2001) A comprehensive two-hybrid analysis to explore the yeast protein interactome. P Natl A Sci 98: 4569-4574. https://doi.org/10.1073/pnas.061034498
|
| [157] |
Ozbabacan SEA, Engin HB, Gursoy A, et al. (2011) Transient protein–protein interactions. Protein Eng Des Sel 24: 635-648. https://doi.org/10.1093/protein/gzr025
|
| [158] |
Morris OM, Torpey JH, Isaacson RL (2021) Intrinsically disordered proteins: Modes of binding with emphasis on disordered domains. Open Biol 11: 210222. https://doi.org/10.1098/rsob.210222
|
| [159] |
Bakker MJ, Svensson O, Sørensen HV, et al. (2024) Exploring the functional landscape of the p53 regulatory domain: the stabilizing role of post-translational modifications. J Chem Theory Comput 20: 5842-5853. https://doi.org/10.1021/acs.jctc.4c00570
|
| [160] |
Hlavacek WS, Faeder JR, Blinov ML, et al. (2003) The complexity of complexes in signal transduction. Biotechnol Bioeng 84: 783-794. https://doi.org/10.1002/bit.10842
|
| [161] |
Johnson S, Hunter T (2005) Kinomics: methods for deciphering the kinome. Nat Methods 2: 17-25. https://doi.org/10.1038/nmeth731
|
| [162] |
Poluri KM, Gulati K, Sarkar S (2021) Experimental methods for determination of protein–protein interactions. Protein-Protein Interactions . Singapore: Springer. https://doi.org/10.1007/978-981-16-1594-8_5
|
| [163] |
Snider J, Kotlyar M, Saraon P, et al. (2015) Fundamentals of protein interaction network mapping. Mol Syst Biol 11: 848. https://doi.org/10.15252/msb.20156351
|
| [164] |
Louche A, Salcedo SP, Bigot S (2017) Protein–protein interactions: pull-down assays. Bacterial Protein Secretion Systems: Methods and Protocols . New York: Springer 247-255. https://doi.org/10.1007/978-1-4939-7033-9_20
|
| [165] |
Brymora A, Valova VA, Robinson PJ (2004) Protein-protein interactions identified by pull-down experiments and mass spectrometry. Curr Protoc Cell Biol 22: 17-25. https://doi.org/10.1002/0471143030.cb1705s22
|
| [166] |
Fuxreiter M (2018) Fuzziness in protein interactions—a historical perspective. J Mol Biol 430: 2278-2287. https://doi.org/10.1016/j.jmb.2018.02.015
|
| [167] |
Gentili PL (2018) The fuzziness of the molecular world and its perspectives. Molecules 23: 2074. https://doi.org/10.3390/molecules23082074
|
| [168] |
Uversky VN (2016) p53 proteoforms and intrinsic disorder: an illustration of the protein structure–function continuum concept. Int J Mol Sci 17: 1874. https://doi.org/10.3390/ijms17111874
|
| [169] | Inae E, Liu G, Jiang M Motif-aware attribute masking for molecular graph pre-training, preprint (2023). https://doi.org/10.48550/arXiv.2309.04589 |
| [170] | Vigo M, Brown J, Conway V (2013) Benchmarking web accessibility evaluation tools: measuring the harm of sole reliance on automated tests. Proceedings of the 10th international cross-disciplinary conference on web accessibility : 1-10. https://doi.org/10.1145/2461121.2461124 |
| [171] |
Rajagopala SV, Uetz P (2011) Analysis of protein–protein interactions using high-throughput yeast two-hybrid screens. Methods Mol Biol 781: 1-29. https://doi.org/10.1007/978-1-61779-276-2_1
|
| [172] | Moore N (2016) Lessons from the fatal French study BIA-10-2474. Bmj 353. https://doi.org/10.1136/bmj.i2727 |
| [173] |
Kaur R, Sidhu P, Singh S (2016) What failed BIA 10–2474 Phase I clinical trial? Global speculations and recommendations for future Phase I trials. J Pharmacol Pharmaco 7: 120-126. https://doi.org/10.4103/0976-500X.189661
|
| [174] |
Burak AT, Turkoglu I (2022) Prediction of viral-host interactions of COVID-19 by computational methods. Chemometr Intell Lab Syst 228: 104622. https://doi.org/10.1016/j.chemolab.2022.104622
|
| [175] |
Iuchi H, Kawasaki J, Kubo K, et al. (2023) Bioinformatics approaches for unveiling virus-host interactions. Comput Struct Biotechnol J 21: 1774-1784. https://doi.org/10.1016/j.csbj.2023.02.044
|
| [176] | Domenech de Cellès M, Goult E, Casalegno JS, et al. (2022) The pitfalls of inferring virus-virus interactions from co-detection prevalence data: application to influenza and SARS-CoV-2. Proc Royal Soc Biol Sci 289: 20212358. https://doi.org/10.1098/rspb.2021.2358 |
| [177] | Alvarez-Ponce D (2017) Recording negative results of protein–protein interaction assays: an easy way to deal with the biases and errors of interactomic data sets. Briefings in Bioinformatics 18: 1017-1020. https://doi.org/10.1093/bib/bbw075 |
| [178] |
Geva G, Sharan R (2010) Identification of protein complexes from co-immunoprecipitation data. Bioinformatics 27: 111-117. https://doi.org/10.1093/bioinformatics/btq652
|
| [179] |
Zhou M, Li Q, Wang PR (2016) Current experimental methods for characterizing protein–protein interactions. Chemmedchem 11: 738. https://doi.org/10.1002/cmdc.201500495
|
| [180] |
Siebenmorgen T, Zacharias M (2020) Computational prediction of protein–protein binding affinities. Wires: Computat Mol Sci 10: e1448. https://doi.org/10.1002/wcms.1448
|
| [181] |
Chaves EJ, Sartori J, Santos WM, et al. (2025) Estimating absolute protein–protein binding free energies by a super learner Model. J Chem Inf Model 65: 2602-2609. https://doi.org/10.1021/acs.jcim.4c01641
|
| [182] |
Pushkaran AC, Arabi AA (2024) From understanding diseases to drug design: can artificial intelligence bridge the gap?. Artif Intell Rev 57: 86. https://doi.org/10.1007/s10462-024-10714-5
|
| [183] |
Wang H (2024) Prediction of protein-ligand binding affinity via deep learning models. Brief Bioinform 25: bbae081. https://doi.org/10.1093/bib/bbae081
|
| [184] |
Wu X, Xu M, Geng M, et al. (2023) Targeting protein modifications in metabolic diseases: molecular mechanisms and targeted therapies. Sig Transduct Target Ther 8: 220. https://doi.org/10.1038/s41392-023-01439-y
|
| [185] |
Hachiya R, Tanaka M, Itoh M, et al. (2022) Molecular mechanism of crosstalk between immune and metabolic systems in metabolic syndrome. Inflamm Regen 42: 13. https://doi.org/10.1186/s41232-022-00198-7
|
| [186] |
Malinverni D, Babu MM (2023) Data-driven design of orthogonal protein-protein interactions. Science signaling 16: eabm4484. https://www.science.org/doi/full/10.1126/scisignal.abm4484
|
| [187] |
Hardy LW, Norton PP (2004) The multiple orthogonal tools approach to define molecular causation in the validation of druggable targets. Drug Discov Today 9: 117-126. https://doi.org/10.1016/S1359-6446(03)02969-6
|
| [188] |
Day ES, Capili AD, Borysenko CW, et al. (2013) Determining the affinity and stoichiometry of interactions between unmodified proteins in solution using Biacore. Anal Biochem 440: 96-107. https://doi.org/10.1016/j.ab.2013.05.012
|
| [189] |
Navarro-Lérida I, Sánchez-Álvarez M, Del Pozo MA (2021) Post-translational modification and subcellular compartmentalization: emerging concepts on the regulation and physiopathological relevance of RhoGTPases. Cells 10: 1990. https://doi.org/10.3390/cells10081990
|
| [190] |
Pan AC, Jacobson D, Yatsenko K, et al. (2019) Atomic-level characterization of protein–protein association. P Natl Acad Sci 116: 4244-4249. https://doi.org/10.1073/pnas.1815431116
|
| [191] |
Mansueto G, Fusco G, Colonna G (2024) A tiny viral protein, SARS-CoV-2-ORF7b: functional molecular mechanisms. Biomolecules 14: 541. https://doi.org/10.3390/biom14050541
|
| [192] | Colonna G A tiny viral protein, SARS-CoV-2-ORF7b: structural features, preprints (2025). https://doi.org/10.20944/preprints202304.0522.v2 |
| [193] |
Whited AM, Johs A (2015) The interactions of peripheral membrane proteins with biological membranes. Chem Phys Lipids 192: 51-59. https://doi.org/10.1016/j.chemphyslip.2015.07.015
|
| [194] |
Oughtred R, Stark C, Breitkreutz BJ, et al. (2019) The BioGRID interaction database: 2019 update. Nucleic Acids Res 47: D529-D541. https://doi.org/10.1093/nar/gky1079
|
| [195] |
Lambert N, Chen YN, Cheng YC, et al. (2013) Quantum biology. Nat Phys 9: 10-18. https://doi.org/10.1038/nphys2474
|
| [196] |
Kim Y, Bertagna F, D'souza EM, et al. (2021) Quantum biology: an update and perspective. Quantum Rep 3: 80-126. https://doi.org/10.3390/quantum3010006
|
| [197] |
Stöhr M, Tkatchenko A (2019) Quantum mechanics of proteins in explicit water: the role of plasmon-like solute-solvent interactions. Sci Adv 5: eaax0024. https://www.science.org/doi/full/10.1126/sciadv.aax0024
|
| [198] |
Layton I, Oppenheim J, Weller-Davies Z (2024) A healthier semi-classical dynamics. Quantum 8: 1565. https://doi.org/10.22331/q-2024-12-16-1565
|
| [199] |
Basieva I, Khrennikov A, Ozawa M (2021) Quantum-like modeling in biology with open quantum systems and instruments. Biosystems 201: 104-328. https://doi.org/10.1016/j.biosystems.2020.104328
|
| [200] | Goh BH, Tong ES, Pusparajah P (2020) Quantum biology: Does quantum physics hold the key to revolutionizing medicine?. Prog Drug Discov Biomed Sci 3: a0000130. https://doi.org/10.36877/pddbs.a0000130 |
| [201] |
Alsteens D (2023) Grand challenges in biophysics. Front Biophys 1: 1215594. https://doi.org/10.3389/frbis.2023.1215594
|
| [202] |
Hatters DM (2023) Grand challenges in biomolecular condensates: structure, function, and formation. Front Biophys 1: 1208763. https://doi.org/10.3389/frbis.2023.1208763
|
| [203] |
Sikosek T, Chan HS (2014) Biophysics of protein evolution and evolutionary protein biophysics. J R Soc Interface 11: 100. https://doi.org/10.1098/rsif.2014.0419
|
| [204] |
Mateus A, Savitski MM, Piazza I (2021) The rise of proteome-wide biophysics. Mol Syst Biol 17: e10442. https://doi.org/10.15252/msb.202110442
|
| [205] |
Echave J, Wilke CO (2017) Biophysical models of protein evolution: understanding the patterns of evolutionary sequence divergence. Annu Rev Biophys 46: 85-103. https://doi.org/10.1146/annurev-biophys-070816-033819
|
| [206] | Zur H, Tuller T (2016) Predictive biophysical modeling and understanding of the dynamics of mRNA translation and its evolution. Nucleic Acids Res 44: 9031-9049. https://doi.org/10.1093/nar/gkw764 |
| [207] |
Skolnick J, Gao M, Zhou H (2016) How special is the biochemical function of native proteins?. F1000Research 5: F1000-Faculty. https://doi.org/10.12688/f1000research.7374.1
|
| [208] |
Yadav R, Gupta DP, Singh C (2025) Lock and key: quest to find the most compatible membrane mimetic for studying membrane proteins in native environment. BBA-Biomembranes 1867: 184414. https://doi.org/10.1016/j.bbamem.2025.184414
|
| [209] |
Staniak M, Huang T, Figueroa-Navedo AM, et al. (2025) Relative quantification of proteins and post-translational modifications in proteomic experiments with shared peptides: a weight-based approach. Bioinformatics 41: btaf046. https://doi.org/10.1093/bioinformatics/btaf046
|
| [210] |
Fumagalli C, Barberis M (2021) Breast cancer heterogeneity. Diagnostics 11: 1555. https://doi.org/10.3390/diagnostics11091555
|
| [211] |
Ribeiro AL, Dallagiovanna B (2025) The role of long non-coding RNAs in human endoderm differentiation. Non-Coding RNA 11: 29. https://doi.org/10.3390/ncrna11020029
|
| [212] |
Costantini S, Sharma A, Raucci R, et al. (2013) Genealogy of an ancient protein family: the Sirtuins, a family of disordered members. BMC Evol Biol 13: 60. https://doi.org/10.1186/1471-2148-13-60
|
| [213] |
Hu KH, Kuhn NF, Courau T, et al. (2023) Transcriptional space-time mapping identifies concerted immune and stromal cell patterns and gene programs in wound healing and cancer. Cell Stem Cell 30: 885-903.e10. https://doi.org/10.1016/j.stem.2023.05.001
|
| [214] |
Droin C, Kholtei JE, Bahar Halpern K, et al. (2021) Space-time logic of liver gene expression at sub-lobular scale. Nat Metab 3: 43-58. https://doi.org/10.1038/s42255-020-00323-1
|
| [215] |
Ross LN (2021) Causal concepts in biology: How pathways differ from mechanisms and why it matters. Brit J Philos Sci 72: 131-158. https://doi.org/10.1093/bjps/axy078
|
| [216] | Bishop RC, Atamanspacher H, Bishop R (2002). Deterministic and indeterministic descriptions, Between Chance and Choice: Interdisciplinary Perspectives on Determinism |